Open Access

Biological functions and clinic significance of SAF‑A (Review)

  • Authors:
    • Daiquan Zhang
    • Li Li
    • Mengni Li
    • Xinmei Cao
  • View Affiliations

  • Published online on: April 10, 2024     https://doi.org/10.3892/br.2024.1776
  • Article Number: 88
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As one member of the heterogeneous ribonucleoprotein (hnRNP) family, scaffold attachment factor A (SAF‑A) or hnRNP U, is an abundant nuclear protein. With RNA and DNA binding activities, SAF‑A has multiple functions. The present review focused on the biological structure and different roles of SAF‑A and SAF‑A‑related diseases. It was found that SAF‑A maintains the higher‑order chromatin organization via RNA and DNA, and regulates transcription at the initiation and elongation stages. In addition to regulating pre‑mRNA splicing, mRNA transportation and stabilization, SAF‑A participates in double‑strand breaks and mitosis repair. Therefore, the aberrant expression and mutation of SAF‑A results in tumors and impaired neurodevelopment. Moreover, SAF‑A may play a role in the anti‑virus system. In conclusion, due to its essential biological functions, SAF‑A may be a valuable clinical prediction factor or therapeutic target. Since the role of SAF‑A in tumors and viral infections may be controversial, more animal experiments and clinical assays are needed.

1. Introduction

Scaffold attachment factor A (SAF-A), or heterogeneous ribonucleoprotein U (hnRNP U), belonging to the hnRNP subfamily, is an abundant component of nuclear matrix and hnRNP particles (1).

The human SAF-A gene, located on chromosome 1 q44, has 2 transcript variants, in which variant 2 lacks a segment in the coding region compared with variant 1(2). Consequently, this causes two isoforms with different lengths, yet the same reading frame.

Since human SAF-A (Uniprot code: Q00839) contains 825 amino acids, its predicted molecular weight is ~90 kDa. But, due to modifications such as phosphorylation, SAF-A is usually observed to be ~120 kDa. SAF-A is widely expressed in various organs or tissues, such as bone marrow, lymphoid tissues, brain, heart, lung and kidney (https://www.proteinatlas.org/ENSG00000153187-HNRNPU). Due to its functional domains, SAF-A binds to both DNA (such as scaffold-attached region DNA) and RNA (such as chromatin-associated RNAs) and plays essential roles in several cellular processes such as chromatin structure regulation (3), transcription (4) and mitosis (5). The present review summarized the structure, multiple functions and clinical significance of SAF-A.

2. Materials and methods

The present narrative review was performed by screening the Pubmed (https://pubmed.ncbi.nlm.nih.gov/), Medline (https://www.nlm.nih.gov/medline/medline_overview.html) and Google Scholar (https://scholar.google.com/) databases using the key words: ‘hnRNPU’, ‘SAF-A’, ‘chromatin organization’, ‘transcription’, ‘RNA metabolism’, ‘DNA repair’, ‘mitosis’, ‘tumor’, ‘neurodevelopment’, and ‘anti-virus’. Published between 2014 and 2024, the English-written articles available with a full text were mainly used, and these articles were involved in animal and human studies.

3. The structure of SAF-A

SAF-A contains three domains: N-terminal DNA-binding, C-terminal RNA binding and middle domain (6-8) (Fig. 1). Notably, the DNA-binding domain SAF/Acinus/PIAS (SAP) is important for interaction with matrix association region (MAR) in chromatin (6). Containing a cluster of arginine/glycine-rich (RGG) repeats, the C-terminus of SAF-A is necessary for binding to inactive X chromosome regions and chromatin-associated RNAs (8). The middle domain of SAF-A, including an SPla and RYanodine receptor and a nucleotide triphosphate hydrolase region (9,10), mediates its interaction with different proteins, such as forkhead box N3(11), Wilms' tumor suppressor gene 1(9) and severe fever with thrombocytopenia syndrome virus nucleocapsid protein (12).

4. Functions of SAF-A

SAF-A participates in chromatin organization

In the eukaryotic cell nucleus, chromatin forms 3-D structures at multiple levels, including domains, loops, A and B compartments (relating to active and inactive chromatin, respectively), topologically associating domains (TADs) and territories (13-16).

In early research, SAF-A was found to form large aggregates in vitro and induce DNA loops to gather around them (17) (Fig. 2A). When SAF-A is depleted in mouse hepatocytes, lamina-associated domains increase, compartments switch, TAD boundary strengthens, and chromatin loop intensities decrease, demonstrating that SAF-A plays a vital role in maintaining the higher-order chromatin organization (18).

Further studies demonstrated that SAF-A regulates chromatin structure via RNA. Jiao et al (19) found that SAF-A binds to histone acetyltransferases p300 in cancer cells (19). Facilitated by heparanase (HPSE) enhancer RNA, the SAF-A-p300 complex is enriched on the super-enhancer, consequently leading to chromatin looping between the HPSE promoter and the super-enhancer. Consistently interacting with chromatin-associated RNAs, SAF-A forms oligomerization that induces de-compaction of large-scale human interphase chromatin structure (8) (Fig. 2A). In this way, SAF-A maintains genomic stability (8). When human monocyte THP-1 cells are infected by vesicular stomatitis virus (VSV), SAF-A interacts with viral infection-induced RNAs, mediating the openness and activation of antiviral immune genes (20). Puvvula and Moon (10) treated cancer cells with cell-penetrating peptides derived from SAF-A, and found that the SAP-derived peptide rather than the RGG-derived one promotes chromatin compaction in HCT116 (colorectal), T47D (breast) and UMUC3 (bladder) cancer cells. Based on these observations, RNA is proposed to be essential for SAF-A-induced chromatin de-compaction.

Besides RNA, DNA was also reported to be necessary for SAF-A to regulate chromatin structure. In the study of Kolpa et al (21), similar to the C280 SAF-A deletion mutant (only containing RGG domain), the expression of ∆RGG (containing DNA-binding domain) and G29A mutants (in lack of DNA-binding ability) was able to release C0T-1 RNA from chromatin, resulting in chromatin condensation. Despite this, their regulatory mechanism is different. ∆RGG SAF-A mutant mainly replaced endogenous SAF-A from chromatin, while the G29A mutant still combined with C0T-1 RNA and could not bind to chromatin (21). Therefore, to some degree, SAF-A may play a role in bridging C0T-1 RNA and chromatin to regulate chromatin architecture. Depending on the SAP domain, SAF-A was also found to combine with MARs and the pericentromere tandem repeats in chromocenters (6,22). Therefore, it was hypothesized that SAF-A tethers MARs to the chromocenter to organize nuclear architecture (1).

SAF-A regulates transcription

In eukaryotic cells, transcription of protein-coding genes, including initiation, elongation and termination phases, contains numerous regulatory proteins affecting either the RNA polymerase II (Pol II) machinery or chromatin structure (23-26).

Previous studies have indicated that SAF-A is involved in initial transcriptional regulation. Actin in the nucleus regulates transcription by remodeling chromatin, regulating transcription factor (TF) location, or binding to RNA polymerase (27-30). Through extracellular vesicles derived from embryonic stem cells, SAF-A is transferred into human coronary artery endothelial cells to combine with actin. The SAF-A-actin complex leads to enhanced RNA Pol II phosphorylation and its level on vascular endothelial growth factor (VEGF) promoter, upregulating VEGF expression (4) (Fig. 2B). When the SAF-A-actin complex is disrupted by H19 [a long non-coding (lnc) RNA], the phosphorylation of the Pol II C-terminal domain (CTD) is inhibited, and consequently, Pol II-mediated transcription is prohibited (31). Additionally, SAF-A was found to associate with elements within the promoter regions. In Sertoli cells, SAF-A has been identified to bind directly to the promoter regions of Sox8 and Sox9, thereby enhancing their expression (32) (Fig. 2B). Similarly, IL21-anti-sense RNA 1 interacting with SAF-A binds to the IL21 promoter, which is essential for regulating IL21 transcription (33). Research on embryonic stem cells also revealed that not only does SAF-A bind to the Oct4 proximal promoter, but it also interacts with endogenous Pol II. And depletion of SAF-A impairs Oct4 expression (34).

In previous studies, SAF-A appeared to have two-way adjusting effects on transcription elongation mediated by Pol II. According to the study of Obrdlik et al (35), SAF-A, actin and p300/CBP-associated factor (PCAF) were associated with the phosphorylated Pol II CTD; and the actin-SAF-A interaction assisted Pol II transcription elongation depending on PCAF (Fig. 2B). On the contrary, in another study, SAF-A inhibited Pol II elongation. Through the middle domain, SAF-A was sufficient to combine with Pol II and repressed TF IIH-mediated Pol II CTD phosphorylation, which inhibited Pol II elongation (36) (Fig. 2B).

SAF-A plays roles in RNA metabolism

SAF-A has also been implicated in various aspects of RNA metabolism, including normal and alternative pre-mRNA splicing and RNA transporting (Fig. 2C).

Ye et al (37) inactivated SAF-A expression in murine hearts and found that compared with wild type, SAF-A mutant in hearts results in extensive intron retention and cassette skipping. In contrast to the wild-type samples, mouse cortices with SAF-A mutations showed 850 differentially spliced genes, and the gene splicing included exon skipping, an alternative 3' or 5' splice site and intron retention (38).

Then, by what mechanism does SAF-A regulate pre-mRNA splicing? Xiao et al (39) found that SAF-A binds to all the small nuclear RNAs essential for splicing major and minor intron classes and regulates U2 small nuclear ribonucleoprotein maturation and Cajal body morphology. However, Vu et al (40) demonstrated the different splicing regulatory mechanisms of SAF-A. Competed with hnRNP L (a splicing repressor) to combine with an RNA cis-element in the exon 3 of caspase-9, SAF-A promotes the exon 3, 4, 5, 6 cassette inclusion in the mature caspase-9 mRNA, determining the preferential expression of caspase-9a rather than caspase-9b.

It has been identified that SAF-A is responsible for transporting and stabilizing mRNA via directly binding to it. Zhao et al (41) found that SAF-A binds to tumor necrosis factor α (TNF-α), IL-6 and IL-1β mRNAs and that downregulation of SAF-A expression in macrophages induces the decreased half-life of these cytokine mRNAs. Besides, Toll-like receptor signaling in macrophages leads to SAF-A translocation from nuclear to cytoplasm (41). During this translocation, SAF-A is proposed to stabilize pro-inflammatory cytokine mRNAs (41). In agreement with the aforementioned study, SAF-A was found to bind to the 3' untranslated region of TNF α mRNA to stabilize it and specifically enhance TNF α expression (42). However, other studies suggested that SAF-A stabilizes mRNA depending on other molecules. Pan et al (43) found that interaction between LIM domains-containing 1 antisense RNA 1 (LIMD1-AS1) and SAF-A is essential for stabilizing LIMD1 mRNA in non-small cell lung cancer. Furthermore, Lu et al (44) reported that following lipopolysaccharide stimulation in human intestinal epithelial cells, functional intergenic repeating RNA element cooperating with SAF-A may regulate the stabilization of vascular cell adhesion protein 1 and IL12p40 mRNAs through targeting the AU-rich elements of these mRNAs.

SAF-A repairs both oxidized lesions and double-strand breaks (DSBs)

In cells, ionizing radiation (IR) and reactive oxygen species may result in clustered oxidized bases and DSBs that are also induced by V(D)J recombination (45-47). Oxidized base lesions in the human genome are repaired via DNA glycosylase (DG) repair, also known as base excision repair (BER). Repair enzymes, including Nei endonuclease VIII-like (NEIL) and certain non-repair proteins, such as SAF-A, initiate this process (48,49). In previous studies, SAF-A was demonstrated to interact directly with NEIL1 and stimulate NEIL1-mediated repair of oxidative base damage (48,49). In addition to NEIL1, SAF-A combines with NEIL2, enhancing NEIL2-initiated transcribed gene-specific repair of oxidized bases (50) (Fig. 2D).

However, BER of bi-stranded oxidized bases can lead to additional DSBs, causing loss of DNA fragments. This may be avoided if DSBs are repaired through non-homologous end joining (NHEJ) mediated by Ku, DNA-dependent protein kinase catalytic subunit (DNA-PKcs), X-ray repair cross-complementing protein 4 (XRCC4), DNA ligase IV, and cernunnos-XRCC4-like factor complex, preceding BER (51-53). In response to irradiation-induced DSBs, SAF-A is rapidly recruited to DNA damage sites (54) (Fig. 2D). Besides, DSBs cause phosphorylated SAF-A at Ser59, which is exclusively dependent on DNA-PK (55,56) (Fig. 2D). In NHEJ-deficient cells treated with IR or clastogenic drugs, the extent and duration of SAF-A phosphorylation at Ser59 increase (56), which suggests that phosphorylated SAF-A may be a biomarker for testing the capacity of the cells to repair DSBs by NHEJ. A previous study showed that SAF-A phosphorylation at Ser59 is related to transient NEIL1 release from chromatin and BER prevention. Then, due to dephosphorylation, SAF-A reactivates BER by relieving DG inhibition (57). These results suggested that DSB repair by NHEJ or BER is partly determined by whether SAF-A is phosphorylated. However, a recent study found that increased SAF-A stabilizes or recruits NHEJ factors via liquid-liquid phase separation, including Ku80, 53BP1, DNA-PKcs and the shieldin complex at DSBs during antibody class-switch recombination (58) (Fig. 2D). The finding supplies SAF-A may balance between BER and NHEL through complex mechanisms.

SAF-A takes part in mitosis

During mitosis, kinetochore-microtubule (MT) attachment and chromosome congression at the spindle equator is essential to accurate chromosome segregation (59,60). Interestingly, SAF-A binds to not only Aurora-A and targeting protein for Xklp2 (TPX2) at spindle MTs, but also nucleolin at the outer kinetochore, whereby SAF-A stabilizes kinetochore-MT attachment (61) (Fig. 2E). Via non-coding RNA, SAF-A interacts with centromere protein W (CENP-W, an inner centromere component crucial for forming a functional kinetochore complex). Notably, SAF-A-CENP-W interaction increases their stability, and they co-localize at the MT-kinetochore interface during mitosis (62) (Fig. 2E). Furthermore, SAF-A depletion in cells leads to delayed mitosis, unsuccessful chromosome alignment, and spindle assembly (61).

In addition, SAF-A is dynamically phosphorylated during mitosis, and in human cells, phosphorylation at serine 2 (S2), S3, S4, S59, S66 and S270 is related to mitosis (63-65). Douglas et al (66) found SAF-A is phosphorylated at S59 by polo-like kinase 1 instead of DNA-PK and is dephosphorylated by protein phosphatase 2A. Mutations of SAF-A S59 in cells lead to aberrant mitoses, including polylobed nuclei, delayed passage, misaligned and lagging chromosomes (66). However, the regulatory mechanism of phosphorylated SAF-A on mitosis remains elusive.

5. Clinical significance of SAF-A

SAF-A is associated with tumor development and resistance to chemotherapy

Previous studies revealed that SAF-A mainly promotes proliferation (67-71), aggressiveness (19) and migration (71) of tumors (Table I). SAF-A is overexpressed in hepatocellular carcinoma or acute myeloid leukemia, and the proliferation of these tumors is inhibited in vitro and in vivo due to SAF-A downregulation (68-70). Further studies demonstrated that SAF-A promotes tumor proliferation through combining with lncRNAs, such as promoter of CDKN1A antisense DNA damage activated RNA (PANDA) in esophageal squamous cell carcinoma and hepatocyte nuclear factor 4 alpha-AS1 in neuroblastoma (67,69). Jiao et al (19) also used several cancer cell lines and found that SAF-A interaction with HPSE enhancer RNA enhances HPSE expression and activity, which promotes tumorigenesis and aggressiveness. In the research of Han et al (71), SAF-A was found to interact with dead box helicase 5 (DDX5) to promote the proliferation and migration of triple-negative breast cancer. On the one hand, the SAF-A-DDX5 complex leads to alternative splicing (AS) of minichromosome maintenance protein 10 pre-mRNA and subsequent activation of Wnt/β-catenin signaling. On the other hand, the complex enhances the expression of LIM domain only protein 4 by locating in its transcriptional start sites, activating PI3K-Akt-mTOR signaling (71). Regulating AS, SAF-A is involved in the pathogenesis of gastric cancer and renal clear cell carcinoma (72,73). Zhang et al (74) found that SAF-A also binds to P-element-induced wimpy testis-interacting RNA-1742 (piRNA-1742) to stabilize USP8 mRNA and promote the progression of renal cell carcinoma. Through interaction with a family with sequence similarity 171 B (an independent prognostic factor for bladder cancer progression), SAF-A regulates the CCL2 pathway to facilitate M2 macrophage polarization in the tumor microenvironment, thereby promoting bladder cancer development (75).

Table I

The roles of SAF-A in various cancers.

Table I

The roles of SAF-A in various cancers.

SystemCancer typeThe roles of SAF-A(Refs.)
DigestiveGastric carcinomaSAF-A is involved in the development of gastric cancer by regulating alternative splicing(72)
 HCCSAF-A promotes HCC development by enhancing CDK2 transcription(68)
 Esophageal squamous cancerLong non-coding RNA PANDA binds to SAF-A to promote tumor proliferation through the CyclinD1/2- Cyclin E1 and Bcl-2 pathways(67)
RespiratoryNon-small cell lung cancer, lung squamous cell carcinomaSAF-A can inhibit tumor proliferation and enhance the sensitivity of lung squamous cell carcinoma to cisplatin(43,78)
Urinary systemBladder carcinomaSAF-A can interact with family with sequence similarity 171 B, an independent predictor of bladder cancer progression, and regulate the CCL2 pathway to promote cancer progression. Knocking down SAF-A can increase the sensitivity of bladder cancer to cisplatin(75,76)
 Renal cell carcinomaA combination of SAF-A and piRNA-1742 stabilizes USP8 mRNA or regulates alternative splicing to promote the progression of renal cell carcinoma(73,74)
Reproductive systemTriple-negative breast cancerInteraction between SAF-A and dead box helicase 5 promotes the proliferation and migration of triple- negative breast cancer(71)
Blood systemMMKnockdown of SAF-A increases MM sensitivity to selinexor(77)

[i] SAF-A, scaffold attachment factor A; HCC, hepatocellular carcinoma; MM, multiple myeloma.

Further studies have demonstrated a correlation between SAF-A and resistance to chemotherapy. Shi et al (76) discovered that the knockout of SAF-A in human T24 cells enhances bladder cancer susceptibility to cisplatin via inhibition of cell proliferation and impairment of cellular invasion and migration capabilities. In the research of Wang et al (77), the knockdown of SAF-A increases selinexor sensitivities of multiple myeloma (MM) cells in vitro and in mouse models and MM patients with relatively low SAF-A expression response to selinexor.

However, a few studies demonstrated that SAF-A might suppress tumor cell proliferation and enhance sensitivities to chemotherapy. Pan et al (43) reported that LIMD1 inhibits proliferation and promotes apoptosis in non-small cell lung cancer cells, and SAF-A interacts with lncRNA LIMD1-AS1 to stabilize LIMD1 mRNA. Puvvula and Moon (10) dealt with various tumor cells with SAF-A RGG- or SAP-derived peptides. Depending on different mechanisms, these peptides suppress breast, bladder, colorectal and prostate cancer cell proliferation. The SAF-A RGG-derived peptides mainly alter SAF-A splicing and binding targets, while the SAP-derived regulates global epigenetic marks to induce DNA damage reaction and cell death (10). Li et al (78) also identified that lncRNA SFTA1 could augment cisplatin sensitivity in treating lung squamous cell carcinoma by upregulating SAF-A.

The contradictory effects of SAF-A on tumors may result from different tumor types or test methods. Therefore, more animal and clinical experiments are needed to identify its functions further.

SAF-A participates in proviral or antiviral response

Previous research revealed that SAF-A plays dual roles in antiviral response. Through the N-terminal fragment (aa1-86), SAF-A targets the 3' long terminal repeat of human immunodeficiency virus type 1 mRNA and blocks viral replication in cells (79). Furtherly, in both in vitro and in vivo animal experiments, SAF-A has been reported to be associated with the initiation of innate immunity against viruses, including severe fever with thrombocytopenia syndrome virus, VSV, and herpes simplex virus type 1 by recognizing nuclear or cytoplasmic viral RNA (12,20,80).

However, Gupta et al (81) found that SAF-A is associated with the leader RNA of VSV and co-localizes with the virus in the cytoplasm of infected cells, implying that SAF-A may contribute to the life cycle of VSV and its pathogenesis. In recent studies, SAF-A was found to negatively regulate innate immune responses against infectious bursal disease viruses and porcine epidemic diarrhea virus and induce immune escape of these viruses (82,83).

Therefore, the diverse roles of SAF-A played in these experiments may be due to different viruses or hosts, and more animal experiments are needed to determine the roles of SAF-A in antiviral immunity.

SAF-A links to neurodevelopment

Caliebe et al (84) first revealed that patients with a chromosome deletion 0.440 Mb in region 1q44 bearing SAF-A suffer from seizures and speech delay. Similarly, Depienne et al (85) reported that 1q43q44 containing SAF-A microdeletion leads to intellectual disability (ID) and epilepsy. Employing molecular genetic testing, a series of pathogenic SAF-A mutants have been identified to be associated with developmental delay and neurodevelopmental disorder showing severe ID with delay of speech and language, early-onset seizures, and autistic features as well (85-90). Despite this, the regulatory mechanism of SAF-A on neurodevelopment remains elusive. In a recent study, deletion of SAF-A in cultured neural progenitors and murine brains resulted in varied gene expression and AS, apoptosis of neural cells and abnormalities in neuronal migration, suggesting that SAF-A is essential for the development of cortex (38).

6. Conclusions

SAF-A is one of the main components of the nuclear matrix. Capable of binding both DNA and RNA, SAF-A has crucial functions in regulating chromatin architecture, transcription, RNA metabolism, DNA repair and mitosis. Humans or mice with SAF-A deficiency or mutants display embryonic death (91), lethal cardiomyopathy (37) and neurodevelopmental disorders (38,90). Therefore, SAF-A may be a valuable prediction factor or therapeutic target. Besides, SAF-A has been observed to be related to anti-infection and tumor progression, though these functions are controversial. Because of this, more animal experiments and clinical assays are needed.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the National Natural Science Foundation of China (grant no. 81801551) and the Central Government Funds of Guiding Local Scientific and Technological Development for Sichuan (grant no. 2021ZYD0076).

Availability of data and materials

Not applicable.

Authors' contributions

XC, ML and DZ contributed to the conception of the study. DZ and LL performed the literature research and wrote the manuscript. XC and ML revised the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Podgornaya OI: Nuclear organization by satellite DNA, SAF-A/hnRNPU and matrix attachment regions. Semin Cell Dev Biol. 128:61–68. 2022.PubMed/NCBI View Article : Google Scholar

2 

Fackelmayer FO and Richter A: hnRNP-U/SAF-A is encoded by two differentially polyadenylated mRNAs in human cells. Biochim Biophys Acta. 1217:232–234. 1994.PubMed/NCBI View Article : Google Scholar

3 

Wavelet-Vermuse C, Odnokoz O, Xue Y, Lu X, Cristofanilli M and Wan Y: CDC20-Mediated hnRNPU ubiquitination regulates chromatin condensation and anti-cancer drug response. Cancers (Basel). 14(3732)2022.PubMed/NCBI View Article : Google Scholar

4 

Wang H, Liu H, Zhao X and Chen X: Heterogeneous nuclear ribonucleoprotein U-actin complex derived from extracellular vesicles facilitates proliferation and migration of human coronary artery endothelial cells by promoting RNA polymerase II transcription. Bioengineered. 13:11469–11486. 2022.PubMed/NCBI View Article : Google Scholar

5 

Sharp JA, Perea-Resa C, Wang W and Blower MD: Cell division requires RNA eviction from condensing chromosomes. J Cell Biol. 219(e201910148)2020.PubMed/NCBI View Article : Google Scholar

6 

Kipp M, Gohring F, Ostendorp T, van Drunen CM, van Driel R, Przybylski M and Fackelmayer FO: SAF-Box, a conserved protein domain that specifically recognizes scaffold attachment region DNA. Mol Cell Biol. 20:7480–7489. 2000.PubMed/NCBI View Article : Google Scholar

7 

Helbig R and Fackelmayer FO: Scaffold attachment factor A (SAF-A) is concentrated in inactive X chromosome territories through its RGG domain. Chromosoma. 112:173–182. 2003.PubMed/NCBI View Article : Google Scholar

8 

Nozawa RS, Boteva L, Soares DC, Naughton C, Dun AR, Buckle A, Ramsahoye B, Bruton PC, Saleeb RS, Arnedo M, et al: SAF-A Regulates Interphase Chromosome Structure through Oligomerization with Chromatin-Associated RNAs. Cell. 169:1214–1227 e18. 2017.PubMed/NCBI View Article : Google Scholar

9 

Spraggon L, Dudnakova T, Slight J, Lustig-Yariv O, Cotterell J, Hastie H and Miles C: hnRNP-U directly interacts with WT1 and modulates WT1 transcriptional activation. Oncogene. 26:1484–1491. 2007.PubMed/NCBI View Article : Google Scholar

10 

Puvvula PK and Moon AM: Novel cell-penetrating peptides derived from scaffold-attachment-factor a inhibits cancer cell proliferation and survival. Front Oncol. 11(621825)2021.PubMed/NCBI View Article : Google Scholar

11 

Zhu X, Huang B, Zhao F, Lian J, He L, Zhang Y, Ji L, Zhang J, Yan X, Zeng T, et al: p38-mediated FOXN3 phosphorylation modulates lung inflammation and injury through the NF-κB signaling pathway. Nucleic Acids Res. 51:2195–2214. 2023.PubMed/NCBI View Article : Google Scholar

12 

Liu BY, Yu XJ and Zhou CM: SAFA initiates innate immunity against cytoplasmic RNA virus SFTSV infection. PLoS Pathog. 17(e1010070)2021.PubMed/NCBI View Article : Google Scholar

13 

Ghirlando R and Felsenfeld G: CTCF: Making the right connections. Genes Dev. 30:881–891. 2016.PubMed/NCBI View Article : Google Scholar

14 

Fritz AJ, Sehgal N, Pliss A, Xu J and Berezney R: Chromosome territories and the global regulation of the genome. Genes Chromosomes Cancer. 58:407–426. 2019.PubMed/NCBI View Article : Google Scholar

15 

Wang W, Chandra A, Goldman N, Yoon S, Ferrari EK, Nguyen SC, Joyce EF and Vahedi G: TCF-1 promotes chromatin interactions across topologically associating domains in T cell progenitors. Nat Immunol. 23:1052–1062. 2022.PubMed/NCBI View Article : Google Scholar

16 

Krasikova A, Kulikova T, Rodriguez Ramos JS and Maslova A: Assignment of the somatic A/B compartments to chromatin domains in giant transcriptionally active lampbrush chromosomes. Epigenetics Chromatin. 16(24)2023.PubMed/NCBI View Article : Google Scholar

17 

Romig H, Fackelmayer FO, Renz A, Ramsperger U and Richter A: Characterization of SAF-A, a novel nuclear DNA binding protein from HeLa cells with high affinity for nuclear matrix/scaffold attachment DNA elements. EMBO J. 11:3431–3440. 1992.PubMed/NCBI View Article : Google Scholar

18 

Fan H, Lv P, Huo X, Wu J, Wang Q, Cheng L, Liu Y, Tang QQ, Zhang L, Zhang F, et al: The nuclear matrix protein HNRNPU maintains 3D genome architecture globally in mouse hepatocytes. Genome Res. 28:192–202. 2018.PubMed/NCBI View Article : Google Scholar

19 

Jiao W, Chen Y, Song H, Li D, Mei H, Yang F, Fang E, Wang X, Huang K, Zheng L and Tong Q: HPSE enhancer RNA promotes cancer progression through driving chromatin looping and regulating hnRNPU/p300/EGR1/HPSE axis. Oncogene. 37:2728–2745. 2018.PubMed/NCBI View Article : Google Scholar

20 

Cao L, Luo Y, Guo X, Liu S, Li S, Li J, Zhang Z, Zhao Y, Zhang Q, Gao F, et al: SAFA facilitates chromatin opening of immune genes through interacting with anti-viral host RNAs. PLoS Pathog. 18(e1010599)2022.PubMed/NCBI View Article : Google Scholar

21 

Kolpa HJ, Creamer KM, Hall LL and Lawrence JB: SAF-A mutants disrupt chromatin structure through dominant negative effects on RNAs associated with chromatin. Mamm Genome. 33:366–381. 2022.PubMed/NCBI View Article : Google Scholar

22 

Lobov IB, Tsutsui K, Mitchell AR and Podgornaya OI: Specificity of SAF-A and lamin B binding in vitro correlates with the satellite DNA bending state. J Cell Biochem. 83:218–229. 2001.PubMed/NCBI View Article : Google Scholar

23 

Cramer P: Organization and regulation of gene transcription. Nature. 573:45–54. 2019.PubMed/NCBI View Article : Google Scholar

24 

Altendorfer E, Mochalova Y and Mayer A: BRD4: A general regulator of transcription elongation. Transcription. 13:70–81. 2022.PubMed/NCBI View Article : Google Scholar

25 

Haberle V and Stark A: Eukaryotic core promoters and the functional basis of transcription initiation. Nat Rev Mol Cell Biol. 19:621–637. 2018.PubMed/NCBI View Article : Google Scholar

26 

Gibbons MD, Fang Y, Spicola AP, Linzer N, Jones SM, Johnson BR, Li L, Xie M and Bungert J: Enhancer-mediated formation of nuclear transcription initiation domains. Int J Mol Sci. 23(9290)2022.PubMed/NCBI View Article : Google Scholar

27 

Sokolova M, Moore HM, Prajapati B, Dopie J, Merilainen L, Honkanen M, Matos RC, Poukkula M, Hietakangas V and Vartiainen MK: Nuclear actin is required for transcription during drosophila oogenesis. iScience. 9:63–70. 2018.PubMed/NCBI View Article : Google Scholar

28 

Knoll KR, Eustermann S, Niebauer V, Oberbeckmann E, Stoehr G, Schall K, Tosi A, Schwarz M, Buchfellner A, Korber P and Hopfner KP: The nuclear actin-containing Arp8 module is a linker DNA sensor driving INO80 chromatin remodeling. Nat Struct Mol Biol. 25:823–832. 2018.PubMed/NCBI View Article : Google Scholar

29 

Almuzzaini B, Sarshad AA, Rahmanto AS, Hansson ML, Von Euler A, Sangfelt O, Visa N, Farrants AK and Percipalle P: In β-actin knockouts, epigenetic reprogramming and rDNA transcription inactivation lead to growth and proliferation defects. FASEB J. 30:2860–2873. 2016.PubMed/NCBI View Article : Google Scholar

30 

Al-Sayegh MA, Mahmood SR, Khair SBA, Xie X, El Gindi M, Kim T, Almansoori A and Percipalle P: beta-actin contributes to open chromatin for activation of the adipogenic pioneer factor CEBPA during transcriptional reprograming. Mol Biol Cell. 31:2511–2521. 2020.PubMed/NCBI View Article : Google Scholar

31 

Bi HS, Yang XY, Yuan JH, Yang F, Xu D, Guo YJ, Zhang L, Zhou CC, Wang F and Sun SH: H19 inhibits RNA polymerase II-mediated transcription by disrupting the hnRNP U-actin complex. Biochim Biophys Acta. 1830:4899–4906. 2013.PubMed/NCBI View Article : Google Scholar

32 

Wen Y, Ma X, Wang X, Wang F, Dong J, Wu Y, Lv C, Liu K, Zhang Y, Zhang Z and Yuan S: hnRNPU in Sertoli cells cooperates with WT1 and is essential for testicular development by modulating transcriptional factors Sox8/9. Theranostics. 11:10030–10046. 2021.PubMed/NCBI View Article : Google Scholar

33 

Liu L, Hu L, Long H, Zheng M, Hu Z, He Y, Gao X, Du P, Zhao H, Yu D, et al: LncRNA IL21-AS1 interacts with hnRNPU protein to promote IL21 overexpression and aberrant differentiation of Tfh cells in systemic lupus erythematosus. Clin Transl Med. 12(e1117)2022.PubMed/NCBI View Article : Google Scholar

34 

Vizlin-Hodzic D, Johansson H, Ryme J, Simonsson T and Simonsson S: SAF-A has a role in transcriptional regulation of Oct4 in ES cells through promoter binding. Cell Reprogram. 13:13–27. 2011.PubMed/NCBI View Article : Google Scholar

35 

Obrdlik A, Kukalev A, Louvet E, Farrants AK, Caputo L and Percipalle P: The histone acetyltransferase PCAF associates with actin and hnRNP U for RNA polymerase II transcription. Mol Cell Biol. 28:6342–6357. 2008.PubMed/NCBI View Article : Google Scholar

36 

Kim MK and Nikodem VM: hnRNP U inhibits carboxy-terminal domain phosphorylation by TFIIH and represses RNA polymerase II elongation. Mol Cell Biol. 19:6833–6844. 1999.PubMed/NCBI View Article : Google Scholar

37 

Ye J, Beetz N, O'Keeffe S, Tapia JC, Macpherson L, Chen WV, Bassel-Duby R, Olson EN and Maniatis T: hnRNP U protein is required for normal pre-mRNA splicing and postnatal heart development and function. Proc Natl Acad Sci USA. 112:E3020–E3029. 2015.PubMed/NCBI View Article : Google Scholar

38 

Sapir T, Kshirsagar A, Gorelik A, Olender T, Porat Z, Scheffer IE, Goldstein DB, Devinsky O and Reiner O: Heterogeneous nuclear ribonucleoprotein U (HNRNPU) safeguards the developing mouse cortex. Nat Commun. 13(4209)2022.PubMed/NCBI View Article : Google Scholar

39 

Xiao R, Tang P, Yang B, Huang J, Zhou Y, Shao C, Li H, Sun H, Zhang Y and Fu XD: Nuclear matrix factor hnRNP U/SAF-A exerts a global control of alternative splicing by regulating U2 snRNP maturation. Mol Cell. 45:656–668. 2012.PubMed/NCBI View Article : Google Scholar

40 

Vu NT, Park MA, Shultz JC, Goehe RW, Hoeferlin LA, Shultz MD, Smith SA, Lynch KW and Chalfant CE: hnRNP U enhances caspase-9 splicing and is modulated by AKT-dependent phosphorylation of hnRNP L. J Biol Chem. 288:8575–8584. 2013.PubMed/NCBI View Article : Google Scholar

41 

Zhao W, Wang L, Zhang M, Wang P, Qi J, Zhang L and Gao C: Nuclear to cytoplasmic translocation of heterogeneous nuclear ribonucleoprotein U enhances TLR-induced proinflammatory cytokine production by stabilizing mRNAs in macrophages. J Immunol. 188:3179–3187. 2012.PubMed/NCBI View Article : Google Scholar

42 

Yugami M, Kabe Y, Yamaguchi Y, Wada T and Handa H: hnRNP-U enhances the expression of specific genes by stabilizing mRNA. FEBS Lett. 581:1–7. 2007.PubMed/NCBI View Article : Google Scholar

43 

Pan J, Tang Y, Liu S, Li L, Yu B, Lu Y and Wang Y: LIMD1-AS1 suppressed non-small cell lung cancer progression through stabilizing LIMD1 mRNA via hnRNP U. Cancer Med. 9:3829–3839. 2020.PubMed/NCBI View Article : Google Scholar

44 

Lu Y, Liu X, Xie M, Liu M, Ye M, Li M, Chen XM, Li X and Zhou R: The NF-κB-Responsive Long Noncoding RNA FIRRE Regulates Posttranscriptional Regulation of Inflammatory Gene Expression through Interacting with hnRNPU. J Immunol. 199:3571–3582. 2017.PubMed/NCBI View Article : Google Scholar

45 

Bredemeyer AL, Helmink BA, Innes CL, Calderon B, McGinnis LM, Mahowald GK, Gapud EJ, Walker LM, Collins JB, Weaver BK, et al: DNA double-strand breaks activate a multi-functional genetic program in developing lymphocytes. Nature. 456:819–823. 2008.PubMed/NCBI View Article : Google Scholar

46 

Cannan WJ, Tsang BP, Wallace SS and Pederson DS: Nucleosomes suppress the formation of double-strand DNA breaks during attempted base excision repair of clustered oxidative damages. J Biol Chem. 289:19881–19893. 2014.PubMed/NCBI View Article : Google Scholar

47 

Nickoloff JA, Sharma N and Taylor L: Clustered DNA Double-Strand Breaks: Biological effects and relevance to cancer radiotherapy. Genes (Basel). 11(99)2020.PubMed/NCBI View Article : Google Scholar

48 

Hegde ML, Banerjee S, Hegde PM, Bellot LJ, Hazra TK, Boldogh I and Mitra S: Enhancement of NEIL1 protein-initiated oxidized DNA base excision repair by heterogeneous nuclear ribonucleoprotein U (hnRNP-U) via direct interaction. J Biol Chem. 287:34202–34211. 2012.PubMed/NCBI View Article : Google Scholar

49 

Dutta A, Yang C, Sengupta S, Mitra S and Hegde ML: New paradigms in the repair of oxidative damage in human genome: Mechanisms ensuring repair of mutagenic base lesions during replication and involvement of accessory proteins. Cell Mol Life Sci. 72:1679–1698. 2015.PubMed/NCBI View Article : Google Scholar

50 

Banerjee D, Mandal SM, Das A, Hegde ML, Das S, Bhakat KK, Boldogh I, Sarkar PS, Mitra S and Hazra TK: Preferential repair of oxidized base damage in the transcribed genes of mammalian cells. J Biol Chem. 286:6006–6016. 2011.PubMed/NCBI View Article : Google Scholar

51 

Hammel M, Yu Y, Radhakrishnan SK, Chokshi C, Tsai MS, Matsumoto Y, Kuzdovich M, Remesh SG, Fang S, Tomkinson AE, et al: An Intrinsically Disordered APLF Links Ku, DNA-PKcs, and XRCC4-DNA Ligase IV in an extended flexible non-homologous end joining complex. J Biol Chem. 291:26987–27006. 2016.PubMed/NCBI View Article : Google Scholar

52 

Weterings E, Verkaik NS, Keijzers G, Florea BI, Wang SY, Ortega LG, Uematsu N, Chen DJ and van Gent DC: The Ku80 carboxy terminus stimulates joining and artemis-mediated processing of DNA ends. Mol Cell Biol. 29:1134–1142. 2009.PubMed/NCBI View Article : Google Scholar

53 

Xing M and Oksenych V: Genetic interaction between DNA repair factors PAXX, XLF, XRCC4 and DNA-PKcs in human cells. FEBS Open Bio. 9:1315–1326. 2019.PubMed/NCBI View Article : Google Scholar

54 

Britton S, Dernoncourt E, Delteil C, Froment C, Schiltz O, Salles B, Frit P and Calsou P: DNA damage triggers SAF-A and RNA biogenesis factors exclusion from chromatin coupled to R-loops removal. Nucleic Acids Res. 42:9047–9062. 2014.PubMed/NCBI View Article : Google Scholar

55 

Berglund FM and Clarke PR: hnRNP-U is a specific DNA-dependent protein kinase substrate phosphorylated in response to DNA double-strand breaks. Biochem Biophys Res Commun. 381:59–64. 2009.PubMed/NCBI View Article : Google Scholar

56 

Britton S, Froment C, Frit P, Monsarrat B, Salles B and Calsou P: Cell nonhomologous end joining capacity controls SAF-A phosphorylation by DNA-PK in response to DNA double-strand breaks inducers. Cell Cycle. 8:3717–3722. 2009.PubMed/NCBI View Article : Google Scholar

57 

Hegde ML, Dutta A, Yang C, Mantha AK, Hegde PM, Pandey A, Sengupta S, Yu Y, Calsou P, Chen D, et al: Scaffold attachment factor A (SAF-A) and Ku temporally regulate repair of radiation-induced clustered genome lesions. Oncotarget. 7:54430–54444. 2016.PubMed/NCBI View Article : Google Scholar

58 

Refaat AM, Nakata M, Husain A, Kosako H, Honjo T and Begum NA: HNRNPU facilitates antibody class-switch recombination through C-NHEJ promotion and R-loop suppression. Cell Rep. 42(112284)2023.PubMed/NCBI View Article : Google Scholar

59 

Tanaka TU: Kinetochore-microtubule interactions: Steps towards bi-orientation. EMBO J. 29:4070–4082. 2010.PubMed/NCBI View Article : Google Scholar

60 

Biggins S and Walczak CF: Captivating capture: How microtubules attach to kinetochores. Curr Biol. 13:R449–R460. 2003.PubMed/NCBI View Article : Google Scholar

61 

Ma N, Matsunaga S, Morimoto A, Sakashita G, Urano T, Uchiyama S and Fukui K: The nuclear scaffold protein SAF-A is required for kinetochore-microtubule attachment and contributes to the targeting of Aurora-A to mitotic spindles. J Cell Sci. 124:394–404. 2011.PubMed/NCBI View Article : Google Scholar

62 

Chun Y, Kim R and Lee S: Centromere Protein (CENP)-W interacts with heterogeneous nuclear ribonucleoprotein (hnRNP) U and may contribute to kinetochore-microtubule attachment in mitotic cells. PLoS One. 11(e0149127)2016.PubMed/NCBI View Article : Google Scholar

63 

Kettenbach AN, Schweppe DK, Faherty BK, Pechenick D, Pletnev AA and Gerber SA: Quantitative phosphoproteomics identifies substrates and functional modules of Aurora and Polo-like kinase activities in mitotic cells. Sci Signal. 4(rs5)2011.PubMed/NCBI View Article : Google Scholar

64 

Malik R, Lenobel R, Santamaria A, Ries A, Nigg EA and Korner R: Quantitative analysis of the human spindle phosphoproteome at distinct mitotic stages. J Proteome Res. 8:4553–4563. 2009.PubMed/NCBI View Article : Google Scholar

65 

Wang Z, Udeshi ND, Slawson C, Compton PD, Sakabe K, Cheung WD, Shabanowitz J, Hunt DF and Hart GW: Extensive crosstalk between O-GlcNAcylation and phosphorylation regulates cytokinesis. Sci Signal. 3(ra2)2010.PubMed/NCBI View Article : Google Scholar

66 

Douglas P, Ye R, Morrice N, Britton S, Trinkle-Mulcahy L and Lees-Miller SP: Phosphorylation of SAF-A/hnRNP-U Serine 59 by Polo-Like Kinase 1 Is Required for Mitosis. Mol Cell Biol. 35:2699–2713. 2015.PubMed/NCBI View Article : Google Scholar

67 

Shi W, Wang Q, Bian Y, Fan Y, Zhou Y, Feng T, Li Z and Cao X: Long noncoding RNA PANDA promotes esophageal squamous carcinoma cell progress by dissociating from NF-YA but interact with SAFA. Pathol Res Pract. 215(152604)2019.PubMed/NCBI View Article : Google Scholar

68 

Liang Y, Fan Y, Liu Y and Fan H: HNRNPU promotes the progression of hepatocellular carcinoma by enhancing CDK2 transcription. Exp Cell Res. 409(112898)2021.PubMed/NCBI View Article : Google Scholar

69 

Song H, Li D, Wang X, Fang E, Yang F, Hu A, Wang J, Guo Y, Liu Y, Li H, et al: HNF4A-AS1/hnRNPU/CTCF axis as a therapeutic target for aerobic glycolysis and neuroblastoma progression. J Hematol Oncol. 13(24)2020.PubMed/NCBI View Article : Google Scholar

70 

Xu CL, Chen Y, Zhu TT, Sun ZJ and Chu JH: Clinical significance and pathogenesis analysis of heterogeneous nuclear ribonucleoprotein U in acute myeloid leukemia. Zhonghua Xue Ye Xue Za Zhi. 43:745–752. 2022.PubMed/NCBI View Article : Google Scholar : (In Chinese).

71 

Han BY, Liu Z, Hu X and Ling H: HNRNPU promotes the progression of triple-negative breast cancer via RNA transcription and alternative splicing mechanisms. Cell Death Dis. 13(940)2022.PubMed/NCBI View Article : Google Scholar

72 

Dong YY, Wang MY, Jing JJ, Wu YJ, Li H, Yuan Y and Sun LP: Alternative Splicing Factor Heterogeneous Nuclear Ribonucleoprotein U as a promising biomarker for gastric cancer risk and prognosis with tumor-promoting properties. Am J Pathol. 194:13–29. 2024.PubMed/NCBI View Article : Google Scholar

73 

Chen T, Zheng W, Chen J, Lin S, Zou Z, Li X and Tan Z: Systematic analysis of survival-associated alternative splicing signatures in clear cell renal cell carcinoma. J Cell Biochem. 121:4074–4084. 2020.PubMed/NCBI View Article : Google Scholar

74 

Zhang W, Zheng Z, Wang K, Mao W, Li X, Wang G, Zhang Y, Huang J, Zhang N, Wu P, et al: piRNA-1742 promotes renal cell carcinoma malignancy by regulating USP8 stability through binding to hnRNPU and thereby inhibiting MUC12 ubiquitination. Exp Mol Med. 55:1258–1271. 2023.PubMed/NCBI View Article : Google Scholar

75 

Hu WM, Li M, Ning JZ, Tang YQ, Song TB, Li LZ, Zou F, Cheng F and Yu WM: FAM171B stabilizes vimentin and enhances CCL2-mediated TAM infiltration to promote bladder cancer progression. J Exp Clin Cancer Res. 42(290)2023.PubMed/NCBI View Article : Google Scholar

76 

Shi ZD, Hao L, Han XX, Wu ZX, Pang K, Dong Y, Qin JX, Wang GY, Zhang XM, Xia T, et al: Targeting HNRNPU to overcome cisplatin resistance in bladder cancer. Mol Cancer. 21(37)2022.PubMed/NCBI View Article : Google Scholar

77 

Wang X, Xu J, Li Q, Zhang Y, Lin Z, Zhai X, Wang F, Huang J, Gao Q, Wen J, et al: RNA-binding protein hnRNPU regulates multiple myeloma resistance to selinexor. Cancer Lett. 580(216486)2024.PubMed/NCBI View Article : Google Scholar

78 

Li L, Yin JY, He FZ, Huang MS, Zhu T, Gao YF, Chen YX, Zhou DB, Chen X, Sun LQ, et al: Long noncoding RNA SFTA1P promoted apoptosis and increased cisplatin chemosensitivity via regulating the hnRNP-U-GADD45A axis in lung squamous cell carcinoma. Oncotarget. 8:97476–97489. 2017.PubMed/NCBI View Article : Google Scholar

79 

Valente ST and Goff SP: Inhibition of HIV-1 gene expression by a fragment of hnRNP U. Mol Cell. 23:597–605. 2006.PubMed/NCBI View Article : Google Scholar

80 

Cao L, Liu S, Li Y, Yang G, Luo Y, Li S, Du H, Zhao Y, Wang D, Chen J, et al: The Nuclear Matrix Protein SAFA Surveils Viral RNA and Facilitates Immunity by Activating Antiviral Enhancers and Super-enhancers. Cell Host Microbe. 26:369–384 e8. 2019.PubMed/NCBI View Article : Google Scholar

81 

Gupta AK, Drazba JA and Banerjee AK: Specific interaction of heterogeneous nuclear ribonucleoprotein particle U with the leader RNA sequence of vesicular stomatitis virus. J Virol. 72:8532–8540. 1998.PubMed/NCBI View Article : Google Scholar

82 

Hu X, Wu X, Ding Z, Chen Z and Wu H: Characterization and functional analysis of chicken dsRNA binding protein hnRNPU. Dev Comp Immunol. 138(104521)2023.PubMed/NCBI View Article : Google Scholar

83 

Zhou H, Yan Y, Gao J, Ma M, Liu Y, Shi X, Zhang Q and Xu X: Heterogeneous Nuclear Protein U Degraded the m(6)A Methylated TRAF3 Transcript by YTHDF2 To Promote Porcine Epidemic Diarrhea Virus Replication. J Virol. 97(e0175122)2023.PubMed/NCBI View Article : Google Scholar

84 

Caliebe A, Kroes HY, van der Smagt JJ, Martin-Subero JI, Tonnies H, van 't Slot R, Nievelstein RA, Muhle H, Stephani U, Alfke K, et al: Four patients with speech delay, seizures and variable corpus callosum thickness sharing a 0.440 Mb deletion in region 1q44 containing the HNRPU gene. Eur J Med Genet. 53:179–185. 2010.PubMed/NCBI View Article : Google Scholar

85 

Depienne C, Nava C, Keren B, Heide S, Rastetter A, Passemard S, Chantot-Bastaraud S, Moutard ML, Agrawal PB, VanNoy G, et al: Genetic and phenotypic dissection of 1q43q44 microdeletion syndrome and neurodevelopmental phenotypes associated with mutations in ZBTB18 and HNRNPU. Hum Genet. 136:463–479. 2017.PubMed/NCBI View Article : Google Scholar

86 

Epi4K Consortium; Epilepsy Phenome/Genome Project. Allen AS, Berkovic SF, Cossette P, Delanty N, Dlugos D, Eichler EE, Epstein MP, Glauser T, et al: De novo mutations in epileptic encephalopathies. Nature. 501:217–221. 2013.PubMed/NCBI View Article : Google Scholar

87 

Shimada S, Oguni H, Otani Y, Nishikawa A, Ito S, Eto K, Nakazawa T, Yamamoto-Shimojima K, Takanashi JI, Nagata S and Yamamoto T: An episode of acute encephalopathy with biphasic seizures and late reduced diffusion followed by hemiplegia and intractable epilepsy observed in a patient with a novel frameshift mutation in HNRNPU. Brain Dev. 40:813–818. 2018.PubMed/NCBI View Article : Google Scholar

88 

Durkin A, Albaba S, Fry AE, Morton JE, Douglas A, Beleza A, Williams D, Volker-Touw CML, Lynch SA, Canham N, et al: Clinical findings of 21 previously unreported probands with HNRNPU-related syndrome and comprehensive literature review. Am J Med Genet A. 182:1637–1654. 2020.PubMed/NCBI View Article : Google Scholar

89 

Wang T, Hoekzema K, Vecchio D, Wu H, Sulovari A, Coe BP, Gillentine MA, Wilfert AB, Perez-Jurado LA, Kvarnung M, et al: Large-scale targeted sequencing identifies risk genes for neurodevelopmental disorders. Nat Commun. 11(4932)2020.PubMed/NCBI View Article : Google Scholar

90 

Taylor J, Spiller M, Ranguin K, Vitobello A, Philippe C, Bruel AL, Cappuccio G, Brunetti-Pierri N, Willems M, Isidor B, et al: Expanding the phenotype of HNRNPU-related neurodevelopmental disorder with emphasis on seizure phenotype and review of literature. Am J Med Genet A. 188:1497–1514. 2022.PubMed/NCBI View Article : Google Scholar

91 

Roshon MJ and Ruley HE: Hypomorphic mutation in hnRNP U results in post-implantation lethality. Transgenic Res. 14:179–192. 2005.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2024
Volume 20 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang D, Li L, Li M and Cao X: Biological functions and clinic significance of SAF‑A (Review). Biomed Rep 20: 88, 2024
APA
Zhang, D., Li, L., Li, M., & Cao, X. (2024). Biological functions and clinic significance of SAF‑A (Review). Biomedical Reports, 20, 88. https://doi.org/10.3892/br.2024.1776
MLA
Zhang, D., Li, L., Li, M., Cao, X."Biological functions and clinic significance of SAF‑A (Review)". Biomedical Reports 20.6 (2024): 88.
Chicago
Zhang, D., Li, L., Li, M., Cao, X."Biological functions and clinic significance of SAF‑A (Review)". Biomedical Reports 20, no. 6 (2024): 88. https://doi.org/10.3892/br.2024.1776